Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Cancer Rep (Hoboken) ; 7(3): e2056, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38522010

RESUMO

BACKGROUND: Immuno-oncology has been focused on T cell-centric approaches until the field recently started appreciating the importance of tumor-reactive antibody production by tumor-infiltrating plasma B cells, and the necessity of developing novel therapeutic antibodies for the treatment of different cancers. RECENT FINDINGS: B lymphocytes often infiltrate solid tumors and the extent of B cell infiltration normally correlates with stronger T cell responses while generating humoral responses against malignant progression by producing tumor antigens-reactive antibodies that bind and coat the tumor cells and promote cytotoxic effector mechanisms, reiterating the fact that the adaptive immune system works by coordinated humoral and cellular immune responses. Isotypes, magnitude, and the effector functions of antibodies produced by the B cells within the tumor environment differ among cancer types. Interestingly, apart from binding with specific tumor antigens, antibodies produced by tumor-infiltrating B cells could bind to some non-specific receptors, peculiarly expressed by cancer cells. Antibody-based immunotherapies have revolutionized the modalities of cancer treatment across the world but are still limited against hematological malignancies and a few types of solid tumor cancers with a restricted number of targets, which necessitates the expansion of the field to have newer effective targeted antibody therapeutics. CONCLUSION: Here, we discuss about recent understanding of the protective spontaneous antitumor humoral responses in human cancers, with an emphasis on the advancement and future perspectives of antibody-based immunotherapies in cancer.


Assuntos
Neoplasias Hematológicas , Neoplasias , Humanos , Neoplasias/terapia , Imunoterapia , Linfócitos B , Antígenos de Neoplasias
2.
Immunity ; 56(11): 2570-2583.e6, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37909039

RESUMO

Dimeric IgA (dIgA) can move through cells via the IgA/IgM polymeric immunoglobulin receptor (PIGR), which is expressed mainly on mucosal epithelia. Here, we studied the ability of dIgA to target commonly mutated cytoplasmic oncodrivers. Mutation-specific dIgA, but not IgG, neutralized KRASG12D within ovarian carcinoma cells and expelled this oncodriver from tumor cells. dIgA binding changed endosomal trafficking of KRASG12D from accumulation in recycling endosomes to aggregation in the early/late endosomes through which dIgA transcytoses. dIgA targeting of KRASG12D abrogated tumor cell proliferation in cell culture assays. In vivo, KRASG12D-specific dIgA1 limited the growth of KRASG12D-mutated ovarian and lung carcinomas in a manner dependent on CD8+ T cells. dIgA specific for IDH1R132H reduced colon cancer growth, demonstrating effective targeting of a cytoplasmic oncodriver not associated with surface receptors. dIgA targeting of KRASG12D restricted tumor growth more effectively than small-molecule KRASG12D inhibitors, supporting the potential of this approach for the treatment of human cancers.


Assuntos
Carcinoma , Imunoglobulina A , Humanos , Imunoglobulina A/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Citoplasma/metabolismo
3.
Blood Adv ; 7(18): 5586-5602, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37531660

RESUMO

The pathogenesis of cutaneous T-cell lymphoma (CTCL) remains unclear. Using single-cell RNA or T-cell receptor (TCR) sequencing of 32 619 CD3+CD4+ and CD26+/CD7+ and 29 932 CD3+CD4+ and CD26-/CD7- lymphocytes from the peripheral blood of 7 patients with CTCL, coupled to single-cell ATAC-sequencing of 26,411 CD3+CD4+ and CD26+/CD7+ and 33 841 CD3+CD4+ and CD26-/CD7- lymphocytes, we show that tumor cells in Sézary syndrome and mycosis fungoides (MF) exhibit different phenotypes and trajectories of differentiation. When compared to MF, Sézary cells exhibit narrower repertoires of TCRs and exhibit clonal enrichment. Surprisingly, we identified ≥200 mutations in hematopoietic stem cells from multiple patients with Sézary syndrome. Mutations in key oncogenes were also present in peripheral Sézary cells, which also showed the hallmarks of recent thymic egression. Together our data suggest that CTCL arises from mutated lymphocyte progenitors that acquire TCRs in the thymus, which complete their malignant transformation in the periphery.


Assuntos
Linfoma Cutâneo de Células T , Micose Fungoide , Síndrome de Sézary , Neoplasias Cutâneas , Humanos , Síndrome de Sézary/genética , Síndrome de Sézary/patologia , Dipeptidil Peptidase 4 , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Micose Fungoide/genética , Micose Fungoide/patologia , Linfoma Cutâneo de Células T/genética , Receptores de Antígenos de Linfócitos T
4.
Nat Commun ; 14(1): 4502, 2023 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-37495577

RESUMO

Interest in spatial omics is on the rise, but generation of highly multiplexed images remains challenging, due to cost, expertise, methodical constraints, and access to technology. An alternative approach is to register collections of whole slide images (WSI), generating spatially aligned datasets. WSI registration is a two-part problem, the first being the alignment itself and the second the application of transformations to huge multi-gigapixel images. To address both challenges, we developed Virtual Alignment of pathoLogy Image Series (VALIS), software which enables generation of highly multiplexed images by aligning any number of brightfield and/or immunofluorescent WSI, the results of which can be saved in the ome.tiff format. Benchmarking using publicly available datasets indicates VALIS provides state-of-the-art accuracy in WSI registration and 3D reconstruction. Leveraging existing open-source software tools, VALIS is written in Python, providing a free, fast, scalable, robust, and easy-to-use pipeline for registering multi-gigapixel WSI, facilitating downstream spatial analyses.


Assuntos
Microscopia , Software , Microscopia/métodos , Tecnologia
5.
Gynecol Oncol ; 173: 114-121, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37121178

RESUMO

OBJECTIVE: To demonstrate that shared antibody responses in endometriosis and endometriosis-associated ovarian cancer spontaneously antagonize malignant progression and can be leveraged to develop future immunotherapies. METHODS: B cells from cyopreserved clear cell ovarian carcinoma (CCC, n = 2), endometrioid ovarian carcinoma (EC, n = 2), and endometriomas (n = 2) were isolated, activated, and EBV-immortalized. Antibodies were purified from B cell supernatants and used for screening arrays containing most of the human proteome. Targets were prioritized based on accessibility (transmembrane or secreted proteins), expression in endometriosis and cancer, and concurrent IgA and IgG responses. We focused on antibodies targeting tumor-promoting syndecan binding protein (SDCBP) to demonstrate anti-tumor activity. Immunoblots and qPCR were performed to assess SDCBP expression in ovarian cancer and endometriosis cell lines and tumor samples. Recombinant IgG4 was generated using the variable heavy and light chains of dominant B cell receptors (BCRs) reacting against the extracellular domain of SDCBP, and used in in vivo studies in human CCC- and high-grade serous ovarian carcinoma (HGSOC)-bearing immunodeficient mice. RESULTS: Nine accessible proteins detected by both IgA and IgG were identified in all samples - including SDCBP, which is expressed in ovarian carcinomas of multiple histologies. Administration of α-SDCBP IgG4 in OVCAR3 (HGSOC), TOV21G and RMG-I (CCC) tumor-bearing mice significantly decreased tumor volume compared to control irrelevant IgG4. CONCLUSIONS: Spontaneous antibody responses exert suboptimal but measurable immune pressure against malignant progression in ovarian carcinomas. Using tumor-derived antibodies for developing novel immunotherapeutics warrants further investigation.


Assuntos
Adenocarcinoma de Células Claras , Carcinoma Endometrioide , Endometriose , Neoplasias Ovarianas , Humanos , Feminino , Animais , Camundongos , Neoplasias Ovarianas/patologia , Apoptose , Formação de Anticorpos , Linhagem Celular Tumoral , Carcinoma Epitelial do Ovário , Carcinoma Endometrioide/patologia , Imunoglobulina A/metabolismo , Adenocarcinoma de Células Claras/patologia , Sinteninas/metabolismo
6.
Mol Cancer Ther ; 21(7): 1184-1194, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35499393

RESUMO

Although chimeric antigen receptor (CAR)-expressing T cells have proven success in hematologic malignancies, their effectiveness in solid tumors has been largely unsuccessful thus far. We found that some olfactory receptors are expressed in a variety of solid tumors of different histologic subtypes, with a limited pattern of expression in normal tissues. Quantification of OR2H1 expression by qRT-PCR and Western blot analysis of 17 normal tissues, 82 ovarian cancers of various histologies, eight non-small cell lung cancers (NSCLCs), and 17 breast cancers demonstrated widespread OR2H1 expression in solid epithelial tumors with expression in normal human tissues limited to the testis. CAR T cells recognizing the extracellular domain of the olfactory receptor OR2H1 were generated with a targeting motif identified through the screening of a phage display library and demonstrated OR2H1-specific cytotoxic killing in vitro and in vivo, using tumor cells with spontaneous expression of variable OR2H1 levels. Importantly, recombinant OR2H1 IgG generated with the VH/VL sequences of the CAR construct specifically detected OR2H1 protein signal in 60 human lung cancers, 40 ovarian carcinomas, and 73 cholangiocarcinomas, at positivity rates comparable with mRNA expression and without OR2H1 staining in 58 normal tissues. CRISPR/Cas9-mediated ablation of OR2H1 confirmed targeting specificity of the CAR and the tumor-promoting role of OR2H1 in glucose metabolism. Therefore, T cells redirected against OR2H1-expressing tumor cells represent a promising therapy against a broad range of epithelial cancers, likely with an admissible toxicity profile.


Assuntos
Neoplasias Pulmonares , Neoplasias Epiteliais e Glandulares , Neoplasias Ovarianas , Receptores Odorantes , Feminino , Humanos , Linhagem Celular Tumoral , Imunoterapia Adotiva , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Receptores Odorantes/metabolismo , Linfócitos T
7.
Cancer Cell ; 40(5): 545-557.e13, 2022 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-35427494

RESUMO

Despite repeated associations between T cell infiltration and outcome, human ovarian cancer remains poorly responsive to immunotherapy. We report that the hallmarks of tumor recognition in ovarian cancer-infiltrating T cells are primarily restricted to tissue-resident memory (TRM) cells. Single-cell RNA/TCR/ATAC sequencing of 83,454 CD3+CD8+CD103+CD69+ TRM cells and immunohistochemistry of 122 high-grade serous ovarian cancers shows that only progenitor (TCF1low) tissue-resident T cells (TRMstem cells), but not recirculating TCF1+ T cells, predict ovarian cancer outcome. TRMstem cells arise from transitional recirculating T cells, which depends on antigen affinity/persistence, resulting in oligoclonal, trogocytic, effector lymphocytes that eventually become exhausted. Therefore, ovarian cancer is indeed an immunogenic disease, but that depends on ∼13% of CD8+ tumor-infiltrating T cells (∼3% of CD8+ clonotypes), which are primed against high-affinity antigens and maintain waves of effector TRM-like cells. Our results define the signature of relevant tumor-reactive T cells in human ovarian cancer, which could be applicable to other tumors with unideal mutational burden.


Assuntos
Memória Imunológica , Neoplasias Ovarianas , Linfócitos T CD8-Positivos , Feminino , Humanos , Linfócitos do Interstício Tumoral , Células T de Memória
8.
Immunity ; 55(1): 115-128.e9, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35021053

RESUMO

The immune checkpoint receptor PD-1 on T follicular helper (Tfh) cells promotes Tfh:B cell interactions and appropriate positioning within tissues. Here, we examined the impact of regulation of PD-1 expression by the genomic organizer SATB1 on Tfh cell differentiation. Vaccination of CD4CreSatb1f/f mice enriched for antigen-specific Tfh cells, and TGF-ß-mediated repression of SATB1 enhanced Tfh differentiation of human T cells. Mechanistically, high Icos expression in Satb1-/- CD4+ T cells promoted Tfh cell differentiation by preventing T follicular regulatory cell skewing and resulted in increased isotype-switched B cell responses in vivo. Ovarian tumors in CD4CreSatb1f/f mice accumulated tumor antigen-specific, LIGHT+CXCL13+IL-21+ Tfh cells and tertiary lymphoid structures (TLS). TLS formation decreased tumor growth in a CD4+ T cell and CXCL13-dependent manner. The transfer of Tfh cells, but not naive CD4+ T cells, induced TLS at tumor beds and decreased tumor growth. Thus, TGF-ß-mediated silencing of Satb1 licenses Tfh cell differentiation, providing insight into the genesis of TLS within tumors.


Assuntos
Centro Germinativo/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Estruturas Linfoides Terciárias/imunologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica , Inativação Gênica , Genótipo , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Fator de Crescimento Transformador beta/genética
9.
Cancer Res ; 82(5): 859-871, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34949671

RESUMO

Recent studies suggest that B cells could play an important role in the tumor microenvironment. However, the role of humoral responses in endometrial cancer remains insufficiently investigated. Using a cohort of 107 patients with different histological subtypes of endometrial carcinoma, we evaluated the role of coordinated humoral and cellular adaptive immune responses in endometrial cancer. Concomitant accumulation of T, B, and plasma cells at tumor beds predicted better survival. However, only B-cell markers corresponded with prolonged survival specifically in high-grade endometrioid type and serous tumors. Immune protection was associated with class-switched IgA and, to a lesser extent, IgG. Expressions of polymeric immunoglobulin receptor (pIgR) by tumor cells and its occupancy by IgA were superior predictors of outcome and correlated with defects in methyl-directed DNA mismatch repair. Mechanistically, pIgR-dependent, antigen-independent IgA occupancy drove activation of inflammatory pathways associated with IFN and TNF signaling in tumor cells, along with apoptotic and endoplasmic reticulum stress pathways, while thwarting DNA repair mechanisms. Together, these findings suggest that coordinated humoral and cellular immune responses, characterized by IgA:pIgR interactions in tumor cells, determine the progression of human endometrial cancer as well as the potential for effective immunotherapies. SIGNIFICANCE: This study provides new insights into the crucial role of humoral immunity in human endometrial cancer, providing a rationale for designing novel immunotherapies against this prevalent malignancy. See related commentary by Osorio and Zamarin, p. 766.


Assuntos
Neoplasias do Endométrio , Receptores de Imunoglobulina Polimérica , Linfócitos B/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Humanos , Imunidade Humoral , Imunoglobulina A/metabolismo , Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/metabolismo , Microambiente Tumoral
10.
Nature ; 591(7850): 464-470, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33536615

RESUMO

Most ovarian cancers are infiltrated by prognostically relevant activated T cells1-3, yet exhibit low response rates to immune checkpoint inhibitors4. Memory B cell and plasma cell infiltrates have previously been associated with better outcomes in ovarian cancer5,6, but the nature and functional relevance of these responses are controversial. Here, using 3 independent cohorts that in total comprise 534 patients with high-grade serous ovarian cancer, we show that robust, protective humoral responses are dominated by the production of polyclonal IgA, which binds to polymeric IgA receptors that are universally expressed on ovarian cancer cells. Notably, tumour B-cell-derived IgA redirects myeloid cells against extracellular oncogenic drivers, which causes tumour cell death. In addition, IgA transcytosis through malignant epithelial cells elicits transcriptional changes that antagonize the RAS pathway and sensitize tumour cells to cytolytic killing by T cells, which also contributes to hindering malignant progression. Thus, tumour-antigen-specific and -antigen-independent IgA responses antagonize the growth of ovarian cancer by governing coordinated tumour cell, T cell and B cell responses. These findings provide a platform for identifying targets that are spontaneously recognized by intratumoural B-cell-derived antibodies, and suggest that immunotherapies that augment B cell responses may be more effective than approaches that focus on T cells, particularly for malignancies that are resistant to checkpoint inhibitors.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoglobulina A/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Linfócitos T Citotóxicos/imunologia , Transcitose , Especificidade de Anticorpos , Antígenos CD/imunologia , Linhagem Celular , Progressão da Doença , Feminino , Humanos , Neoplasias Ovarianas/prevenção & controle , Receptores Fc/imunologia , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Transcitose/imunologia , Microambiente Tumoral/imunologia
11.
J Clin Invest ; 131(3)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33270606

RESUMO

Cutaneous T cell lymphoma (CTCL) has a poorly understood etiology and no known cure. Using conditional knockout mice, we found that ablation of the genomic organizer special AT-rich sequence-binding protein 1 (Satb1) caused malignant transformation of mature, skin-homing, Notch-activated CD4+ and CD8+ T cells into progressively fatal lymphoma. Mechanistically, Satb1 restrained Stat5 phosphorylation and the expression of skin-homing chemokine receptors in mature T cells. Notably, methyltransferase-dependent epigenetic repression of SATB1 was universally found in human Sézary syndrome, but not in other peripheral T cell malignancies. H3K27 and H3K9 trimethylation occluded the SATB1 promoter in Sézary cells, while inhibition of SUV39H1/2 methyltransferases (unlike EZH2 inhibition) restored protective SATB1 expression and selectively abrogated the growth of primary Sézary cells more effectively than romidepsin. Therefore, inhibition of methyltransferases that silence SATB1 could address an unmet need for patients with mycosis fungoides/Sézary syndrome, a set of incurable diseases.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Metiltransferases/antagonistas & inibidores , Proteínas de Neoplasias , Síndrome de Sézary/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Linhagem Celular Tumoral , Humanos , Metiltransferases/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Síndrome de Sézary/metabolismo , Síndrome de Sézary/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
12.
Science ; 369(6506): 942-949, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32820120

RESUMO

Gamma delta (γδ) T cells infiltrate most human tumors, but current immunotherapies fail to exploit their in situ major histocompatibility complex-independent tumoricidal potential. Activation of γδ T cells can be elicited by butyrophilin and butyrophilin-like molecules that are structurally similar to the immunosuppressive B7 family members, yet how they regulate and coordinate αß and γδ T cell responses remains unknown. Here, we report that the butyrophilin BTN3A1 inhibits tumor-reactive αß T cell receptor activation by preventing segregation of N-glycosylated CD45 from the immune synapse. Notably, CD277-specific antibodies elicit coordinated restoration of αß T cell effector activity and BTN2A1-dependent γδ lymphocyte cytotoxicity against BTN3A1+ cancer cells, abrogating malignant progression. Targeting BTN3A1 therefore orchestrates cooperative killing of established tumors by αß and γδ T cells and may present a treatment strategy for tumors resistant to existing immunotherapies.


Assuntos
Antígenos CD/imunologia , Butirofilinas/antagonistas & inibidores , Butirofilinas/imunologia , Linfócitos Intraepiteliais/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/terapia , Animais , Anticorpos Monoclonais/uso terapêutico , Antígenos CD/genética , Butirofilinas/genética , Feminino , Humanos , Imunoterapia/métodos , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Immunol ; 203(12): 3447-3460, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31704881

RESUMO

Tumor-associated macrophages are major contributors to malignant progression and resistance to immunotherapy, but the mechanisms governing their differentiation from immature myeloid precursors remain incompletely understood. In this study, we demonstrate that exosomes secreted by human and mouse tumor-educated mesenchymal stem cells (MSCs) drive accelerated breast cancer progression by inducing differentiation of monocytic myeloid-derived suppressor cells into highly immunosuppressive M2-polarized macrophages at tumor beds. Mechanistically, MSC-derived exosomes but not exosomes from tumor cells contain TGF-ß, C1q, and semaphorins, which promote myeloid tolerogenic activity by driving PD-L1 overexpression in both immature myelomonocytic precursors and committed CD206+ macrophages and by inducing differentiation of MHC class II+ macrophages with enhanced l-Arginase activity and IL-10 secretion at tumor beds. Accordingly, administration of tumor-associated murine MSC-derived exosomes accelerates tumor growth by dampening antitumor immunity, and macrophage depletion eliminates exosome-dependent differences in malignant progression. Our results unveil a new role for MSC-derived exosomes in the differentiation of myeloid-derived suppressor cells into macrophages, which governs malignant growth.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Exossomos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células Mieloides/metabolismo , Animais , Biomarcadores Tumorais , Neoplasias da Mama/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Modelos Animais de Doenças , Transição Epitelial-Mesenquimal , Feminino , Xenoenxertos , Humanos , Imunomodulação , Imunofenotipagem , Ativação de Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Células Mieloides/citologia
14.
Br J Cancer ; 121(8): 679-689, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31481734

RESUMO

BACKGROUND: Cancer-associated fibroblasts (CAFs) are known to impact on tumour behaviour, but the mechanisms controlling this are poorly understood. METHODS: Breast normal fibroblasts (NFs) or CAFs were isolated from cancers by laser microdissection or were cultured. Fibroblasts were transfected to manipulate miR-222 or Lamin B receptor (LBR). The fibroblast-conditioned medium was collected and used to treat epithelial BC lines MDA-MB-231 and MDA-MB-157. Migration, invasion, proliferation or senescence was assessed using transwell, MTT or X-gal assays, respectively. RESULTS: MiR-222 was upregulated in CAFs as compared with NFs. Ectopic miR-222 expression in NFs induced CAF-like expression profiles, while miR-222 knockdown in CAFs inhibited CAF phenotypes. LBR was identified as a direct miR-222 target, and was functionally relevant since LBR knockdown phenocopied miR-222 overexpression and LBR overexpression phenocopied miR-222 knockdown. MiR-222 overexpression, or LBR knockdown, was sufficient to induce NFs to show the CAF characteristics of enhanced migration, invasion and senescence, and furthermore, the conditioned medium from these fibroblasts induced increased BC cell migration and invasion. The reverse manipulations in CAFs inhibited these behaviours in fibroblasts, and inhibited paracrine influences on BC cells. CONCLUSION: MiR-222/LBR have key roles in controlling pro-progression influences of CAFs in BC. This pathway may present therapeutic opportunities to inhibit CAF-induced cancer progression.


Assuntos
Neoplasias da Mama/genética , Fibroblastos Associados a Câncer/metabolismo , Carcinoma/genética , MicroRNAs/genética , Receptores Citoplasmáticos e Nucleares/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Técnicas de Reprogramação Celular , Senescência Celular , Meios de Cultivo Condicionados , Feminino , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Microdissecção e Captura a Laser , Invasividade Neoplásica , Metástase Neoplásica , Receptor de Lamina B
15.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 502-511, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30553016

RESUMO

Lethal metastasis of primary breast tumors to lymph nodes has been found to be associated with the co-expression of chemokine CXCL13 and its receptor CXCR5. To date, however, the precise molecular events regulating the co-expression of CXCL13 and CXCR5 in the context of breast cancer progression have not been identified. Therefore, to extend our understanding of the drivers of breast cancer metastasis, we undertook a line of investigation in this study in which we demonstrate that the transcriptional regulation of CXCL13 is mediated by the reciprocal activity of RelA and Nrf2, while CXCR5 is transcriptionally silenced by CpG island methylation within its promoter. Critically, we show that intra-tumoral CXCL13 and CXCR5 mRNA expression is positively correlated with intra-tumoral RelA expression within the primary tumor of breast cancer (BCa) patients (n = 98). We demonstrate a role for Nrf2 in the negative transcriptional regulation of cxcl13. Furthermore, using a luciferase assay and deletion analysis of the cxcl13 gene promoter, we demonstrate that RelA and Nrf2 directly act upon the cxcl13 promoter to regulate transcription. Chromatin immunoprecipitation PCR, supported by in silico docking analyses, confirmed that RelA and Nrf2 both occupy multiple positions within the cxcl13 promoter. Collectively, in RelA high conditions, low Nrf2 and lack of cxcr5 promoter DNA-methylation govern CXCL13-CXCR5 co-expression within breast tumors, and thus drive disease progression and metastasis.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Quimiocina CXCL13/genética , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Receptores CXCR5/genética , Fator de Transcrição RelA/metabolismo , Transcrição Gênica , Adulto , Idoso , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Quimiocina CXCL13/metabolismo , Metilação de DNA/genética , Feminino , Humanos , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Fator 2 Relacionado a NF-E2/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR5/metabolismo , Adulto Jovem
16.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2053-2066, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29526821

RESUMO

The multifunctional cytokine TGF-ß crucially participates in breast cancer (BCa) metastasis and works differently in the disease stages, thus contributing in BCa progression. We address connections between TGF-ß and the stem cell-related transcription factor (TF) Oct4 in BCa. In 147 BCa patients with infiltrating duct carcinoma, we identified a significantly higher number of cases with both moderate/high Oct4 expression and high TGF-ß in late stages compared to early stages of the disease. In vitro studies showed that TGF-ß elevated Oct4 expression, which in turn, regulated Epithelial-to-Mesenchymal transition (EMT)-regulatory gene (Snail and Slug) expression, migratory ability, chemotactic invasiveness and extracellular matrix (ECM) degradation potential of BCa cells. Putative binding sites for Oct4 on the snail, slug and cxcl13 promoters and for Smad3 on the snail and slug promoters were identified. Promoter activities of snail and slug were greater in dual-treated cells than only TGF-ß-treated or Oct4-overexpressing cells. CXCL13 mRNA fold changes, however, were low in cells induced with TGF-ß, compared to dual-treated or Oct4-overexpressing cells. Our co-IP studies confirmed that Oct4 and Smad3 form heterodimers that recognize specific promoter sequences to promote Snail and Slug expression, but which in turn, indirectly inhibits Smad3-mediated repression of CXCL13 expression, allowing Oct4 to act as a positive TF for CXCL13. Taken together, these data suggest that TGF-ß signaling and Oct4 cooperate to induce expression of EMT-related genes Snail, Slug and CXCL13, which accelerates disease progression, particularly in the late stages, and may indicate a poor prognosis for BCa patients.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Fator 3 de Transcrição de Octâmero/metabolismo , Proteína Smad3/metabolismo , Fatores de Transcrição da Família Snail/genética , Adulto , Idoso , Mama/patologia , Neoplasias da Mama/genética , Carcinoma Ductal de Mama/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Biologia Computacional , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Regiões Promotoras Genéticas , Multimerização Proteica , Transdução de Sinais/genética , Fatores de Transcrição da Família Snail/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
17.
Immunobiology ; 223(2): 151-161, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29107385

RESUMO

We investigated expressions of -CC chemokine ligand 2 (CCL2) and CCL5 in tumor samples from 147 breast cancer (BCa) patients and correlated with transforming growth factor-ß (TGF-ß) expression. We observed an inverse correlation of TGF-ß expression with CCL2, CCL5 expression in early stages of BCa. On contrary, in late stages, CCL2, not CCL5, expression was found to be directly proportional with TGF-ß expression. TGF-ß stimulated MDA-MB-231 cells to express CCL2, however, downregulated both CCL2 and CCL5 in MCF-7. Interestingly, a significant swing of Th1-Th2 ratio towards Th2 is seen within the primary tumors expressing moderate/high-CCL2-low/negative-CCL5. We observed that CCL2-CCR2 interaction induces monocytes/macrophages to secrete Th2-attracting chemokine CCL22 in vitro. Therefore, CCL2 secreted from the tumor microenvironment may attract and interact with monocytes/macrophages, and favor Th2 accumulation by inducing CCL22 secretion. Study in 4T1-BALB/c BCa mouse model demonstrated significant (p<0.05) decrease in CCL2, CCL5 and CCL22 levels and reduction in lung metastatic nodule numbers upon administering TGF-ß inhibitor. These findings collectively indicate that TGF-ß regulates CCL2 and CCL5 expression in a stage-dependent manner during BCa progression, which in turn, determines Th1-Th2 balance within the tumor microenvironment.


Assuntos
Neoplasias da Mama/imunologia , Quimiocina CCL2/metabolismo , Quimiocina CCL5/metabolismo , Neoplasias Pulmonares/imunologia , Macrófagos/imunologia , Células Th2/imunologia , Animais , Neoplasias da Mama/patologia , Carcinogênese , Quimiocina CCL2/genética , Quimiocina CCL22/metabolismo , Quimiocina CCL5/genética , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Neoplasias Pulmonares/secundário , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Equilíbrio Th1-Th2 , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
18.
J Photochem Photobiol B ; 175: 269-281, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28923599

RESUMO

Silver nanoparticles (AgNPs) have been synthesized in situ in micelles formed by the bile salt sodium deoxycholate (NaDC). The AgNPs exhibit "green" fluorescence. It has been shown in the present study that they can disrupt the components of gall stones/pigment stones. This unique ability of the AgNPs has been observed upon detailed study of the interaction between the endobiotic pigment bilirubin (BR) and bile salt (NaDC). In addition, these AgNPs show significant cytotoxicity towards the breast cancer cells (MCF-7). Thus the AgNPs synthesized in this work show important physiological activity and can serve as prospective "Theranostic Materials" in future. Their green fluorescence bears relevance to future diagnostic applications while their anticancer activity and disruptive action upon BR aggregates in bile salt micelles is extremely important for therapeutic purpose. This is the first report of the use of metal nanoparticles in disruption of components of gall stones/pigment stones and thus the present work has very important physiological significance. The detailed spectral studies indicate that bile salts increase the dimerization of BR which could be linked to increased solubilisation of BR in bile salt media and consequent bile stone/pigment stone formation. Importantly, an increase in red fluorescence was observed (upon dimerization of BR), which is important for cancer detection and studying the metabolism of biological tissues.


Assuntos
Antineoplásicos/química , Ácidos e Sais Biliares/química , Nanopartículas Metálicas/química , Pigmentos Biológicos/química , Prata/química , Antineoplásicos/farmacologia , Bilirrubina/química , Ligação Competitiva , Pontos de Checagem do Ciclo Celular , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Difusão Dinâmica da Luz , Humanos , Células MCF-7 , Nanopartículas Metálicas/toxicidade , Micelas , Microscopia Eletrônica de Transmissão , Pigmentos Biológicos/metabolismo , Espectrometria de Fluorescência , Nanomedicina Teranóstica
20.
Breast Cancer Res Treat ; 143(2): 265-76, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24337540

RESUMO

We investigated the expression of -CXC chemokine ligand 13 (CXCL13) and its receptor -CXC chemokine receptor 5 (CXCR5) in 98 breast cancer (BC) patients with infiltrating duct carcinoma, out of which 56 were found lymph node metastasis (LNM) positive. Interestingly, co-expression of CXCL13 and CXCR5 showed a significant correlation with LNM. Since, epithelial to mesenchymal transition (EMT) is highly associated with metastasis we investigated EMT-inducing potential of CXCL13 in BC cell lines. In CXCL13-stimulated BC cells, expression of various mesenchymal markers (Vimentin, N-cadherin), EMT regulators (Snail, Slug), and matrix metalloproteinase-9 (MMP9) was increased, whereas the expression of epithelial marker E-cadherin was found to be decreased. In addition, expression of receptor activator of nuclear factor kappa-B ligand (RANKL), which is known to regulate MMP9 expression via Src activation, was also significantly increased after CXCL13 stimulation. Using specific protein kinase inhibitors, we confirmed that CXCL13 stimulated EMT and MMP9 expression via RANKL-Src axis in BC cell lines. To further validate this observation, we examined gene expression patterns in primary breast tumors and detected significantly higher expression of various mesenchymal markers and regulators in CXCL13-CXCR5 co-expressing patients. Therefore, this study showed the EMT-inducing potential of CXCL13 as well as demonstrated the prognostic value of CXCL13-CXCR5 co-expression in primary BC. Moreover, CXCL13-CXCR5-RANKL-Src axis may present a therapeutic target in LNM positive BC patients.


Assuntos
Neoplasias da Mama/patologia , Quimiocina CXCL13/metabolismo , Transição Epitelial-Mesenquimal , Metástase Linfática/patologia , Receptores CXCR5/metabolismo , Adulto , Idoso , Antígenos CD/biossíntese , Biomarcadores Tumorais/metabolismo , Caderinas/biossíntese , Linhagem Celular Tumoral , Movimento Celular , Quimiocina CXCL13/antagonistas & inibidores , Quimiocina CXCL13/biossíntese , Feminino , Furanos/farmacologia , Humanos , Indóis/farmacologia , Metaloproteinase 9 da Matriz/biossíntese , Pessoa de Meia-Idade , Inibidores de Fosfoinositídeo-3 Quinase , Piridinas/farmacologia , Pirimidinas/farmacologia , Ligante RANK/biossíntese , Ligante RANK/genética , RNA Mensageiro/biossíntese , Receptores CXCR5/antagonistas & inibidores , Receptores CXCR5/biossíntese , Transdução de Sinais , Fatores de Transcrição da Família Snail , Sulfonamidas/farmacologia , Fatores de Transcrição/biossíntese , Vimentina/biossíntese , Quinases da Família src/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...